Open Access

Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions

  • Authors:
    • Yuehan Wang
    • Mingxia Xu
    • Yeli Yao
    • Ying Li
    • Songfa Zhang
    • Yunfeng Fu
    • Xinyu Wang
  • View Affiliations

  • Published online on: February 22, 2024     https://doi.org/10.3892/ol.2024.14300
  • Article Number: 167
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor invasion and metastasis are the processes that primarily cause adverse outcomes in patients with cervical cancer. Cancer‑associated fibroblasts (CAFs), which participate in cancer progression and metastasis, are novel targets for the treatment of tumors. The present study aimed to assess the heterogeneity of CAFs in the cervical cancer microenvironment through single‑cell RNA sequencing. After collecting five cervical cancer samples and obtaining the CAF‑associated gene sets, the CAFs in the cervical cancer microenvironment were divided into myofibroblastic CAFs and extracellular (ec)CAFs. The ecCAFs appeared with more robust pro‑tumorigenic effects than myCAFs according to enrichment analysis. Subsequently, through combining the ecCAF hub genes and bulk gene expression data for cervical cancer obtained from The Cancer Genome Atlas and Gene Ontology databases, univariate Cox regression and least absolute shrinkage and selection operator analyses were performed to establish a CAF‑associated risk signature for patients with cancer. The established risk signature demonstrated a stable and strong prognostic capability in both the training and validation cohorts. Subsequently, the association between the risk signature and clinical data was evaluated, and a nomogram to facilitate clinical application was established. The risk score was demonstrated to be associated with both the tumor immune microenvironment and the therapeutic responses. Moreover, the signature also has predictive value for the prognosis of head and neck squamous cell carcinoma, and bladder urothelial carcinoma, which were also associated with human papillomavirus infection. In conclusion, the present study assessed the heterogeneity of CAFs in the cervical cancer microenvironment, and a subgroup of CAFs that may be closely associated with tumor progression was defined. Moreover, a signature based on the hub genes of ecCAFs was shown to have biomarker functionality in terms of predicting survival rates, and therefore this CAF subgroup may become a therapeutic target for cervical cancer in the future.

Introduction

Cervical cancer is the fourth most frequently diagnosed cancer and the fourth leading cause of cancer-related mortality in women worldwide (1). Based on the Global Cancer Statistics resource, there were ~604,000 new cases of cervical cancer and 342,000 deaths resulting from cervical cancer worldwide in 2020, and the mortality rate was considerably higher in developing countries compared with developed countries (1). The main pathogenic factor of cervical cancer is high-risk human papillomavirus (HPV)-persistent infection, which has reportedly led to >90% of cervical cancer cases (2). With the application of early screening and diagnostic methods, a larger number of patients can receive a favorable prognosis; however, for advanced, metastatic and recurrent cervical cancer, the prognosis is not optimistic. Based on clinical findings, the 12-month survival rate of patients with advanced cervical cancer has never exceeded 30%, and the response rate is <15% (3). In recent years, immunotherapy and tumor-targeting therapies have yielded promising results for the treatment of solid tumors. However, unlike other solid tumors, there are few targeted drugs available at present for the treatment of cervical cancer, and the treatment options for advanced cervical cancer remains very limited (4,5). Therefore, the identification of novel therapeutic targets for the treatment of cervical cancer is urgently needed.

As a central component of the tumor microenvironment (TME) in solid tumors, the pro-tumorigenic functions of cancer-associated fibroblasts (CAFs) have been assessed in breast cancer, colorectal cancer and numerous other cancer types (6). CAFs are typically associated with a worse prognosis and are currently being studied as a potential therapeutic target (7). However, CAFs also exhibit a certain degree of versatility, which may present certain difficulties in terms of translating research findings into clinical practice. The current classic classification includes myofibroblastic CAFs, which are associated with smooth muscle contraction and collagen precipitation, immune regulatory CAFs, which are associated with immune cell recruitment and suppression, and antigen presenting CAFs, which could recruit T cells, and the composition of CAFs is different in different tumors (8). In cervical cancer, fibroblasts have been reported to be more sensitive to HPV-infected keratinocytes and to secrete fibroblast growth factors (FGFs) such as FGF-2 and FGF-7, through the paracrine system, which in turn promotes angiogenesis and tumor proliferation (9). Therefore, an improved understanding of the biology of CAFs may lead to prognostic and therapeutic benefits.

Based on the potential of single-cell (sc)RNA-sequencing (seq) to decipher cellular heterogeneity, the present study aimed to assess the intra-tumor heterogeneity of CAFs in cervical cancer using this method to further examine CAF function. Through the validation of the findings using multiple databases and clinical data, the preliminary results in the present study may be useful as a reference point for further research.

Materials and methods

Participants and sample collection

Clinical data and tissue specimens were collected from patients with cervical cancer treated at the Women's Hospital, School of Medicine, Zhejiang University (Hangzhou, China) between June 2020 and December 2021. The inclusion criteria were as follows: i) Adult female patients with a sexual history; ii) diagnosis of squamous cervical cancer; iii) no neoadjuvant therapy before surgery; and iv) Federation of Gynecology and Obstetrics (FIGO) 2018 stage ≥IB1 (10). The exclusion criteria were as follows: i) Patients not diagnosed with primary squamous cervical cancer; ii) no lesions visible to the naked eye; and iii) any treatments, such as radiotherapy and chemotherapy, received before surgery. Patients were considered to have cervical cancer according to preoperative histological and/or imaging examinations, such as ultrasound, magnetic resonance imaging or computed tomography.

Tumor samples of ~0.5×0.5×0.5 cm were collected from within the visible tumor area during surgery. Para-tumorous tissues, which were defined as normal tissues according to the naked eye and located 2 cm away from the cancer margin, were also collected if available. The size of the sample collected was the same as that for the tumor tissues. After the sampling was completed, blood and debris on the surface of the tissue were cleaned with normal saline that had been pre-cooled at 4°C. Liquid on the surface of the tissue was then absorbed by dust-free paper and the tissue samples were transferred to a frozen tube filled with the pre-cooled tissue preservation solution (MACS® Tissue Storage Solution; cat. no. 130-100-008; Miltenyi Biotec, Inc.). The tube cover was tightened, sealed with sealing film, and then stored and transported at 2–8°C. The fresh tissues collected were processed within 24 h to prepare for the following steps. Before the final diagnosis was made according to the postoperative pathology and if the patient met the inclusion and exclusion criteria, the sequencing data would be used for subsequent analysis.

In total, five tumor and three para-tumorous tissues were obtained (median patient age, 63 years; range, 48–67 years). The cancer clinical stage was defined according to the aforementioned FIGO 2018 staging system, and stages I, II and III were all included in the present study. As the samples were collected at different times, batch effects were not introduced when analyzing the scRNA-seq data.

The protocol of the present study was approved by the Institutional Research Ethics Committee of the Women's Hospital, School of Medicine, Zhejiang University (approval no. IRB-20200006-R). All patients provided written informed consent. The clinical characteristics of the patients and samples, including age, histological type and presence of metastasis, are detailed in Table SI.

Cell suspensions from tumor tissues and cell quality control

Fresh cervical cancer tissue samples were placed in a sterile RNase-free culture dish containing 3–5 ml calcium-free and magnesium-free 1X PBS on ice. The tissue samples were transferred to new culture dishes and cut into 0.5 mm2 pieces. The tissues were subsequently washed with 1X PBS, and as much irrelevant tissue as possible, including blood stains and fatty layers, was removed. The tissues were then dissociated into single cells using dissociation solution (0.35% collagenase I–V5, 2 mg/ml papain, 120 U/ml DNase I) in a water bath at 37°C with shaking for 20 min (100 rev/min). The digestion was terminated by the addition of 1X PBS containing 10% fetal bovine serum (v/v), and the solution was then resuspended 5–10 times using a Pasteur pipette. The resulting cell suspension was filtered with a 70–30 µm stacked cell strainer and then centrifuged at 300 × g for 5 min at 4°C (11). The cell pellet was subsequently resuspended in 100 µl 1X PBS containing 0.04% BSA, to which 1 ml 1X red blood cell lysis buffer [Red Blood Cell Lysis Solution (10X); cat. no. 130-094-183; Miltenyi Biotec, Inc.] was added. The cell suspension was then incubated either at room temperature or on ice for 2–10 min to lyse the remaining red blood cells. Following this incubation, the cell suspension was centrifuged at 300 × g for 5 min at room temperature. The cells were subsequently resuspended in 100 µl Dead Cell Removal MicroBeads, and dead cells were removed using the Dead Cell Removal Kit (cat. no. 130-090-101; Miltenyi Biotec, Inc.). The cells were then resuspended in 1X PBS containing 0.04% BSA, centrifuged at 300 × g for 3 min at 4°C (this process was repeated twice), before 50 µl 1X PBS containing 0.04% BSA was again added to resuspend the cell pellet. The overall cell viability was determined by trypan blue exclusion staining, and cell viability >85% was required for subsequent experiments. The single-cell suspensions were counted using a Countess® II Automated Cell Counter (Invitrogen; Themo Fisher Scientific, Inc.), and the cell concentration was adjusted to 700–1,200 cells/µl.

10X Genomics library preparation and scRNA-seq

Gel beads containing the barcode information (16-base sequence) were bound to a mixture of the cells to sequence and enzymes (including reverse transcriptase), and then encapsulated in droplets of oil surfactant located in a microfluidic system to form the gel beads in emulsion (GEMs). The GEMs were subsequently collected in to a reservoir in the equipment called the chromium controller, and then reverse transcription was performed in a PCR instrument (Bio-Rad Laboratories, Inc.) to generate cDNA. In this process, the gel beads were lysed during heating and released the barcode sequences that could label the sample cells. At the same time, there were also a large amount of primer sequences in gel beads. The protocol for this step was 53°C for 45 min, 85°C for 5 min and a 4°C hold. The GEMs were then collected, and the cDNA was used as a template for PCR amplification. Subsequently, the GEMs were disrupted and broken up into oil droplets. The cDNA was then enzymatically digested into fragments of 200–300 bp, and the DNA library was finally amplified by PCR following the library construction process that comprised traditional second-generation sequencing, including sequencing connectors and primers. The reagents used in this process were as follows: i) Chromium Next GEM Single Cell 3′ GEM, Library & Gel Bead Kit v3.1, 16 rxns PN-1000121 (10X Genomics); and ii) Chromium Next GEM Chip G Single Cell Kit, 48 rxns PN-1000120 (10X Genomics). Subsequently, an Agilent 2100 bioanalyzer (Agilent Technologies, Inc.) was used for cDNA library quality control. When the peak shape was normally distributed, between 500 and 8,000 bp, and the main peak was >1,300 bp, follow-up experiments could be conducted. The cDNA library was quantitatively determined by qPCR using VAHTS Library Quantification Kit for Illumina (Vazyme NQ101, NQ105) to the concentration of 150 pM. Finally, libraries were sequenced using a NovaSeq 6000 Sequencing System at a minimum depth of 20,000 reads per cell (12) (paired-end multiplexing run; 150 bop; Illumina, Inc.) by using NovaSeq 6000 S4 Reagent Kit v1.5 (300 cycles) (cat. no. 20028312). These experiments which including cDNA library construction and single-cell sequencing were supported by LC-Bio Technologies Hangzhou Co., Ltd., and single-cell sequencing related reagents and instruments were from 10X Genomics.

Sequencing data quality control and quantification

Cell Ranger (4.0.0) is the official analysis software of 10X Genomics (https://support.10×genomics.com/single-cell-gene expression/software/overview/welcome) (13), which is capable of reading raw downstream sequencing data directly and can output the statistical results of the sequencing data for each sample. As the present study involved multiple samples, integration and homogenization of the multi sample data was required prior to further analysis, and to obtain uniform UMI abundance information for all the genes in all cells. After running the whole process, Cell Ranger used algorithms to calculate the number of cells obtained during the process based on the barcode and UMI information, and the expression level of genes was thereby detected in each cell, providing data that were used for further downstream analyses.

Single cell data filtering and cluster analysis

The Cell Ranger output was loaded into the Seurat (version 3.1.1) (http://seurat.r-forge.r-project.org) program for dimensional reduction, clustering and analysis of the scRNA-seq data. The quality control threshold parameters were set as follows: i) Genes that were expressed in <300 cells were removed; ii) the low cut-off was >500 genes expressed per cell, whereas <5,000 genes expressed per cell was the high cut-off; iii) the number of UMI counts was ≥500; iv) the proportion of mitochondrial DNA-derived genes expressed was <25%; and v) the potential doublets that occurred in the encapsulation step and/or as occasional pairs of cells were removed using the Doublet Finder package (version 2.0.2) (https://rdrr.io/github/chris-mcginnis-ucsf/DoubletFinder/src/R/doubletFinder.R).

After the cell filtering process was completed, the Seurat software was used to normalize the data and to find highly variable genes using the built-in parameters ‘Normalize Data’ and ‘Find Variable Features’. Seurat was then used to perform a downstream principal component (PC) analysis (PCA) on these highly variable genes calculated by ‘Find Variable Features’ to derive the corresponding PCA values, and subsequently the data were analyzed according to these values for dimensionality reduction, clustering and subgrouping. The first 20 PCs of the PCA were then used for the subsequent clustering and subgrouping analysis.

To visualize the data, the dimensionality of all 33,090 cells from five tumor samples was further reduced using Seurat, and the non-linear dimensional reduction technique, t-distributed Stochastic Neighbor Embedding, was used to project the cells into two-dimensional (2D) space. The principle was to drop the expression matrix data of the cells onto a 2D plane by means of dimensionality reduction, and to differentiate the data according to similarity to obtain the statistical results of the cell clustering.

Finding marker genes based on the clustering of subgroups

To find clusters, the weighted Shared Nearest Neighbor graph-based clustering method was selected. Marker genes for each cluster were identified using the ‘Find All Markers’ function in Seurat (14) and the Wilcoxon rank sum test was performed (default parameter: ‘bimod’ Likelihood-ratio test). This function allowed the pre-processed data to be limited to a certain range, thus eliminating undesirable effects caused by exceptionally large or small datasets. The following were parameters selected for the genes: i) Genes expressed in >10% of the cells in a cluster; ii) adjusted P<0.01; and iii) an average log2 fold change (FC) of >0.26, which implied a fold change of >1.3. Due to the large amount of single-cell data, this difference was already very notable. Typically, logFC >0.25 is acceptable in single-cell analysis; however, the criteria were made more stringent in the present study to increase the reliability (1517). Subsequently, cell clusters in the resulting 2D representation were annotated to known biological cell types using canonical marker genes that have been reported in previously published studies (1820). Finally, marker genes of the clusters were revealed by heatmap analysis, and heatmaps were generated using the pheatmap function (1.0.12) from the R package (R Core Team). Furthermore, when comparing differential genes of two groups of cells, the basic principles of statistics was similar with finding marker genes of several clusters and the function in Seurat was called ‘Find Marker’.

Pathway and functional annotation analysis

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway functional enrichment analyses were performed using the Cluster Profiler R package (version 4.0.5), through which enriched biological processes, molecular functions, cellular components and pathways of identified hub genes in the cluster of interest were identified (9). P<0.05 was considered to indicate a statistically significant enrichment (21). The results of the enrichment analysis are shown as scatterplots, and the ggplot2:Rich factor was used to indicate the number of differential genes/total number of genes, with the greater the Rich factor, the higher the degree of enrichment.

The Molecular Signatures Database (https://www.gsea-msigdb.org/gsea/msigdb) (genesets c2.cp.kegg.v7.0.symbols and h.all.v7.0.symbols) was used in conjunction with the fgsea R package (version 1.18.0) for gene set enrichment analysis (GSEA) (22,23) to assess the different KEGG pathways and hallmark gene sets between the high- and low-risk CAF groups. Pathways with adjusted P<0.05 were regarded as being statistically significantly enriched (18). The results of the GSEA were visualized using the enrichplot R package (version 1.12.3).

Cell-cell communication analysis

Cell communication analysis was performed after 1,000 permutation tests based on gene expression levels of the major cell types, and the cell-cell interactions among them were visualized by heatmaps using CellPhoneDB (version 2.1.2; www.cellphonedb.org), a publicly available repository of curated receptors, ligands and their interactions. Cell-cell interactions within identical cellular lineages were excluded, and receptor-ligand pairs between the relevant cell types were identified. A combined P<0.05 was required, and the ligand/receptor needed to have been expressed in >10% of cells (24).

Database bulk transcriptome RNA-seq data collection and processing

The bulk transcriptome RNA-seq data and corresponding clinical data were obtained from The Cancer Genome Atlas (TCGA) (https://portal.gdc.cancer.gov) cervical cancer (CESC; 309 samples), head and neck squamous cancer (HNSC; 546 samples), bladder cancer (BLCA; 430 samples), colon adenocarcinoma (COAD; 512 samples) and prostate cancer (PRAD; 551 samples) databases using the UCSC Xena browser (GDC hub; http://gdc.xenahubs.net) (25). Additionally, the transcriptomic data of 300 cervical cancer samples in GSE44001 were obtained as the external validation cohorts via the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/geo/) (26).

Molecular prognostic signature construction

Focusing on the hub genes of extracellular marked CAFs and differentially expressed genes (DEGs) between CAFs and fibroblasts in para-tumors, a prognostic signature was constructed for patients with cervical cancer. Overall survival (OS) rate was identified as the primary outcome. First, univariate Cox regression analysis was performed to identify prognostic ecCAF-associated genes in TCGA-CESC dataset. P<0.05 was considered to indicate a statistically significant result. To minimize the overfitting risk, the least absolute shrinkage and selection operator (LASSO) Cox regression model was then applied using the glmnet 4.1 4 R package (27), and the CAF signature was calculated according to the following formula: CAF risk score=Σ(βi × Expi), where βi represents the LASSO coefficient of the gene and Expi is the expression value of the candidate gene (28). Patients in TCGA-CESC database were classified into high- and low-risk groups according to the median CAF risk score, and their association with OS was evaluated using Kaplan-Meier (KM) plot analysis (29). Heatmap analysis was used to visualize the association between CAF risk scores and candidate genes. Similarly, the CAF signature was tested in other external validation cohorts, including the aforementioned cervical cancer cohort GSE44001 and other HPV-associated cancer datasets, including TCGA-HNSC, TCGA-BLCA, TCGA-COAD and TCGA-PRAD datasets (21).

TME infiltration estimation

The Estimation of Stromal and Immune cells in Malignant Tumors using Expression data (ESTIMATE) algorithm in R package (version 1.0.13) was used to calculate the immune scores and tumor purity of the patients. In addition, the Wilcoxon rank-sum test was used to compare immune scores and tumor purity between the CAF high- and low-risk score groups (30).

The infiltration levels of different immune cells between the CAF high- and low-risk score groups were calculated using the following two methods: i) The combination of Cell type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) via ‘CIBERSORT.R’ and LM22 (leukocyte signature matrix) was used to evaluate the proportions of the 22 human leukocyte cell subsets; and ii) single-sample (ss)GSEA analysis was applied via the GSVA R package (version 1.42.0) (31) to assess the proportions of 28 types of infiltrating immune cells of the tumor samples (32). Subsequently, the Wilcoxon rank-sum test was used to compare the differences between the two groups, and the ggplot R package was used for visualization of the results.

Immunotherapeutic responses and chemotherapeutic sensitivity predictions

The immunotherapeutic responses of patients in TCGA-CESC database and other cancer databases were estimated using the online algorithm, Tumor Immune Dysfunction and Exclusion (TIDE; http://tide.dfci.harvard.edu/) (33). Furthermore, patient chemotherapeutic sensitivities were predicted using the oncoPredict R package (version 0.2) by building a ridge regression model (34). Several common anticancer drugs for cervical cancer (cisplatin, docetaxel, 5-fluorouracil, paclitaxel and cyclophosphamide) were tested, and the training genetic profiles were obtained from the largest publicly accessible pharmacogenomics database, Genomics of Drug Sensitivity in Cancer (GDSC; http://www.cancerrxgene.org/) (21,35). GDSC contains two datasets, the newer dataset, GDSC2, was used in the present study.

Nomogram construction and validation

To identify which clinical characteristics were associated with the prognosis of patients with cervical cancer, univariate and multivariate Cox proportional hazards regression analyses were performed using SPSS 26.0 (IBM Corp.). After passing the Schoenfeld residual plot of proportional hazards test, the significant variables were tested using the proportional hazards assumption (36). Nomograms were then constructed based on the independent prognostic factors in the Cox regression to predict the 1-, 3- and 5-year OS rates of patients with cervical cancer (37). Every variable in the nomogram was expressed as a line, and the length of the lines reflected the weight of the variables in the model; the value of the variable was equal to a point on the line. After adding the points of all variables, the survival rates at different time nodes were obtained (21).

In addition, the concordance index (C-index) was calculated to evaluate the level of discrimination of the model, and calibration curves were plotted to assess the consistency between the predicted 1-, 3- and 5-year OS rate probabilities and the observed rates, the accuracies of which were derived from the nomogram (bootstrap-based 1,000 iterations, with resampling of validations) (38).

Human Protein Atlas (HPA) database

Immunohistochemical results were obtained from the HPA online database (https://www.proteinatlas.org/) (39), to compare the expression levels of the signature genes at the protein level in normal cervix and cervical cancer tissues.

Statistical analysis

All statistical analyses and visualizations were performed using the R v4.1.1 (https://www.r-project.org/) and SPSS 26.0 software packages. KM survival curves and the log-rank test were used to compare the OS rates between high- and low-risk groups, and these analyses were performed using survival and survminer R packages (40). Furthermore, the survival results of only one gene were obtained from the Gene Expression Profiling Interactive Analysis database (http://gepia.cancer-pku.cn). The LASSO-Cox regression model was used to construct the extracellular (ec)CAF signature. Time-dependent receiver operating characteristic (ROC) curves were used to evaluate the predictive performance of the model. Wilcoxon and unpaired Student's t-tests were used to compare differences between two groups, depending on whether the data were normally distributed. χ2 tests were used to compare constituent ratios between two groups. Finally, Pearson's correlation coefficient was used to assess correlations between two variates. P<0.05 was considered to indicate a statistically significant difference.

Results

scRNA-seq analysis and CAF subgroup identification

A flow diagram depicting the overall steps of the present study is presented in Fig. 1. A total of five fresh tumor tissues and three available para-tumorous tissues were collected from patients with cervical cancer, and scRNA-seq was subsequently performed with all samples. Following quality control assessment and the removal of batch effects, a total of 33,090 single cells from tumor samples were sorted into 20 major clusters (Figs. 2A and S1A). Heatmap analysis was then conducted to reveal the top 10 hub genes of the 20 clusters (Fig. 2B), and a bar-plot was constructed to demonstrate the sample source of the clusters (Fig. 2C). Subsequently, the cell types were annotated by finding canonical markers in cluster-specific genes (Fig. 2D): Epithelial cellular adhesion molecule was used for identifying epithelial cells (Fig. 2E); protein tyrosine phosphatase, receptor type C was used for identifying immune cells (Fig. 2F); claudin-5 was used for endothelial cells (Fig. 2G); and collagen, type I, α1 was used for CAFs. Two clusters, namely cluster 10 and cluster 16 (C10 and C16, respectively), were recognized as CAFs (Fig. 2H).

Subsequently, the heterogeneity of CAFs in a cervical cancer environment was evaluated by assessing the gene expression profile of the two clusters. One difference identified was in the expression of α-smooth muscle actin (α-SMA; also known as ACTA2), a classical marker of myofibroblastic (my)CAFs (41). ACTA2 was expressed at notable levels in C16, whereas the expression level in C10 was comparatively low (Fig. 2E). With the exception of ACTA2, C16 also exhibited notably higher expression levels of other intracellular markers, including vimentin and ferroptosis suppressor protein 1 (also known as S100A4) compared with C10; whereas C10 expressed extracellular markers, including lumican (LUM), matrix metalloproteinase (MMP)11, MMP2 and fibulin 1 (FBLN1) (Fig. 2H). Previous studies have suggested that MMPs remodel the extracellular matrix (ECM) via the proteolysis of collagens, fibrin, fibronectin, laminins and vitronectin, and through promotion of the invasion of tumor cells in an HPV-infected microenvironment (42,43). For these reasons, the C10 CAF subgroup was classified as ecCAFs in the present study. In addition, certain genes that are characteristic of CAF subpopulations that have been reported in other types of tumor were also assessed, including immune regulatory CAFs and antigen-presenting CAFs (8). However, these types of CAFs did not appear to exist in the cervical cancer microenvironment, as both of the clusters did not express classical marker,s such as IL6, CXCL1, CXCL12, HLA-DRA and HLA-DPA1 (Fig. 2H).

Subsequently, cell-cell communication analysis was performed to assess the interactions among different cell types in the cervical cancer microenvironment (Fig. 2I). The results demonstrated that ecCAFs had a notably higher number of interactions with other cell types compared with myCAFs (C16), as a greater number of ligand-receptor pairings were detected. The majority of clusters that made connections with ecCAFs were cancer cells (clusters 3, 4, 5, 6, 7, 13, 14, 15 and 17; cancer cells originated from epithelial cells that highly express EPCAM), followed by monocytes (cluster 8, which was associated with a high expression level of CD14). This suggested that, when compared with myCAFs, ecCAFs may more notably contribute towards promoting cancer progression.

ecCAFs demonstrate more robust pro-tumorigenic effects compared with myCAFs

As myCAFs are the major subtype of activated fibroblasts in the TME, they are thought to have a pro-tumorigenic role (8). myCAFs were therefore used as the reference cells in the present study, to assess the function of ecCAFs in the cervical cancer microenvironment. First, the DEGs were compared between the two subgroups. Volcano plots were constructed to demonstrate the most significantly up and downregulated genes in ecCAFs compared with myCAFs (P<0.01; Fig. 3A). In addition to LUM and MMP11, numerous other genes that fulfill a crucial role in the ECM were found to be upregulated, including decorin (44), FBLN1 (45), MMP2, neuroblastoma 1 and spondin 2 (46), whereas genes that serve regulatory roles in certain intercellular signaling pathways and mitochondrial functions, such as NADH dehydrogenase 1 α subcomplex, 4 like 2 (47) and neurogenic locus notch homolog protein 3 (48), were downregulated. Moreover, genes that control myogenesis such as myosin heavy chain 11 (49) and myocyte-specific enhancer factor 2C (50), were also demonstrated to be upregulated in myCAFs and downregulated in ecCAFs.

GO and KEGG enrichment analyses were then performed for the DEGs identified between ecCAFs and myCAFs. In the GO enrichment analysis, the DEGs were found to be closely associated with ECM organization (Fig. 3B). KEGG enrichment analysis revealed that, even though the two types of CAFs were both associated with cancer, ecCAFs were more abundant in cancer-linked signaling pathways and cancer-associated components (P<0.05; Fig. 3C), such as ‘PI3K-Akt signaling pathway’, ‘TGF-beta signaling pathway’, ‘Pathways in cancer’, ‘Proteoglycans in cancer’, ‘Transcriptional misregulation in cancer’ and ‘MicroRNAs in cancer’. These findings corroborated that the novel extracellular marker CAFs, namely ecCAFs, had a more significant role in cervical cancer progression compared with the classic intracellular marker CAFs (or myCAFs). Moreover, the ECM may be an important location for ecCAFs in terms of their support of tumor progression, as focal adhesion and ECM-receptor interactions were also found to be significantly enriched (Fig. 3C and D). Notably, the two subgroups of CAFs were also demonstrated to be significantly associated with HPV infection.

Construction of a prognostic signature based on the hub genes of ecCAFs

Given the cancer-promoting functions of ecCAFs, ecCAFs were subsequently used to predict the prognosis of patients with cervical cancer. TCGA cervical cancer cohort, which contained 296 samples, was used to construct the model, and the OS rate was chosen as the primary outcome. Both the hub genes of ecCAFs and the differential gene profiles of CAFs in cervical cancer tissues and fibroblasts in para-tumorous tissues on the single-cell level were taken into consideration. Finally, 51 intersecting genes, which were hub genes only in ecCAFs when compared with myCAFs, and fibroblasts in paratumor were obtained. Univariate Cox proportional risk models were then used to analyze the intersecting genes, and 16 genes were identified (P<0.05; Fig. 4A). The LASSO Cox regression algorithm was then performed on these genes, and the λmin values (namely, the value of λ that gave the minimum mean cross-validated error) was determined as the optimal λ value by tenfold cross validations (Fig. 4B). A total of 12 prognostic genes with non-zero coefficients were identified. A 12-gene ecCAF signature was subsequently constructed based on the expression level of each gene, and its coefficient was as follows: Risk score=[0.228614961999191 × heparan sulfate proteoglycan 2 (HSPG2)] + [0.280769639745156 × leptin receptor (LEPR) overlapping transcript (OT)] + (0.164309473932873 × LEPR)-[(0.101889575739853 × LIM domain only 4 (LMO4)]-[(0.0924809661366454 × vascular cell adhesion molecule 1 (VCAM1)]-[(0.120237842213106 × regulator of G protein signaling 5 (RGS5)] + [(0.0176198675612701 × prostaglandin endoperoxide synthase 2 (PTGS2)] + [(0.398174149944109 × transmembrane protein 9 (TMEM9)] + [(0.0569947847625145 × collagen, type IV, α1 (COL4A1)]-[(0.11168153683065 × NMYC downstream-regulated gene 2 (NDRG2)] + [(0.0959648255524675 × Egl 9 family hypoxia inducible factor 3 (EGLN3)] + [(0.0896858331705779 × SEC23 homolog A, COPII coat complex component (SEC23A)]. Among the prognostic genes, eight genes, HSPG2, LEPROT, LEPR, PTGS2, TMEM9, COL4A1, EGLN3 and SEC23A, were regarded as risk-associated genes (HR>1), whereas the other four genes were considered as protective genes (HR <1). Fig. 4C shows the change in survival status (dead/alive) of patients in the TCGA-CESC cohort with the increase of risk score, and the patients are divided into high risk and low risk groups by median. Based on the aforementioned risk formula, the ecCAF risk score of each patient was calculated and visualized by heatmap analysis (Fig. 4D). Patients in TCGA-CESC cohort were subsequently divided into low and high ecCAF risk groups according to their median risk scores. The Wilcoxon rank-sum test demonstrated that the ecCAF risk score in the group of dead patients was significantly higher compared with the group of live patients (P=3×10−8; Fig. 4E). The pairwise comparison of OS in the different risk groups was then assessed using the log-rank test. KM curves revealed that the high ecCAF risk group had a significantly more unfavorable survival outcome compared with the low ecCAF risk group (P<0.001; Fig. 4F). ROC curves were subsequently used to evaluate the sensitivity and specificity of the OS model. The overall area under the ROC curve (AUC) value was found to be 0.724 (Fig. 4G), and for the 1-, 3- and 5-year survival prognoses, the constructed risk model predicted AUC values of 79.92, 79.26 and 81.68, respectively (Fig. 4H).

Subsequently, the reliability of the ecCAF signature was further assessed. When building this signature, the OS rate was defined as the primary outcome; however, when changing the survival outcome to other parameters, including to disease-specific survival, disease free interval and progression free interval, the model also demonstrated robustness in terms of its forecast performance when dividing the patients according to the median ecCAF risk score (P<0.01; Fig. 5A-C). Cervical cancer information is limited in the public datasets selected, and therefore the GSE44001 dataset, which contained information regarding cancer recurrence, was selected as the validation dataset. The signature was also able to predict the prognosis of patients when the cut-off point was set at the upper quartile of the ecCAF risk score (P=0.012; Fig. 5D). Furthermore, the survival results of every model gene were obtained from the Gene Expression Profiling Interactive Analysis database. The survival results revealed that 7/12 genes (COL4A1, EGLN3, LEPR, LEPROT, SEC23A, TMEM9 and VCAM1) had prognostic functions that were consistent with the risk model results of the present study when the cut-off point was set at the median value (Fig. S2). Patients with higher VCAM1 expression levels had an improved prognosis, whereas a higher expression level of other genes indicated a worse prognosis for survival. Taken together, these results supported the prediction for the OS rate of patients with CESC according to the constructed model.

Furthermore, to evaluate the protein expression levels of the model genes in normal and tumor fibroblasts, immunohistochemical results were obtained from the HPA database. In total, 6/12 genes (SEC23A, NDRG2, PTGS2, VCAM1, LMO4 and HSPG2) were found to have immunohistochemical results, and SEC23A, VCAM1, LMO4 and HSPG2 were found to be expressed at a notably higher level in cervical cancer stroma compared with normal cervical stroma (Fig. 5E). These findings were consistent with the results of single cell sequencing showing that the expression of these genes was upregulated in ecCAFs compared with that in fibroblasts in para-tumor tissues. By contrast, there was no notable difference in the protein expression levels of NDRG2 and PTGS2 between the tumor and normal groups in HPA.

Higher ecCAF risk indicates an immune exclusive environment and does not respond well to immunotherapy or chemotherapy

The influence of the ecCAF risk score on the TME was evaluated by ESTIMATE (51). In TCGA-CESC cohort, patients with a higher ecCAF risk score exhibited significantly higher tumor purity (Wilcoxon rank-sum test, P=0.003; Fig. 6B) and lower immune cell infiltration (Wilcoxon rank-sum test, P<0.001; Fig. 6A), compared with patients with a lower risk score. Subsequently, CIBERSORT and the ssGSEA algorithms were used to assess the relationship between the ecCAF risk score and TME constituents at the bulk RNA-seq level. The results demonstrated that a high ecCAF risk score was significantly associated with a higher level of macrophages 0, which represents the initial state of the macrophage, compared with a low-risk score, whereas there was a deficit of a majority of immune contents, including several subtypes of B cells, CD4 T cells, CD8 T cells and dendritic cells, all of which usually have antitumor functions in the tumor microenviroment (5254) (Fig. 6C-E). In conclusion, a higher ecCAF risk score may indicate a microenvironment that is more exclusive to immune cells.

Since it was determined that the ecCAF risk score may influence the TME outlook, particularly in terms of immune cells, the practicability of the model in predicting the response to immunotherapy in cervical cancer was evaluated. According to the Pearson's correlation coefficient analysis, the ecCAF signature score was significantly but weakly negatively correlated with the expression levels of programmed cell death protein 1 (P<0.001; r=−0.23; Fig. 7A) and cytotoxic T-lymphocyte associated protein 4 (P<0.001; r=−0.29; Fig. 7B). The probability of samples in the CESC cohorts yielding a response to immune-checkpoint inhibitors was predicted using the TIDE online algorithm. Patients in the low ecCAF risk group (148/296; non-responders, n=24) were significantly more reactive to immunotherapy compared with patients in the high-ecCAF risk group (148/296; non-responders, n=38) (χ2 test, P<0.05; Fig. 7C). In addition, the non-responders had a significantly higher ecCAF risk score compared with responders (Student's t-test, P<0.01; Fig. 7D). These results demonstrated that the constructed model could be used to provide an indication as to whether a patient may be an immunotherapy responder.

Subsequently, the ability of the constructed signature to predict response to chemotherapy was evaluated. The oncoPredict algorithm (36) was used to estimate the sensitivity of five frequently used drugs in cervical cancer therapy (cisplatin, docetaxel, 5-fluorouracil, paclitaxel and cyclophosphamide), and a training cohort was obtained from the GDSC2 dataset (a resource for therapeutic biomarker discovery in cancer cells). Of all five drugs assessed, docetaxel and paclitaxel were significantly associated with improved therapeutic effects compared with the other drugs, as their sensitivity scores were relatively small. In addition, the samples with a higher ecCAF risk score had significantly lower sensitivity than those with a lower risk score (Wilcoxon rank-sum test, all P<0.05; Fig. 7E-I).

Clinical applications of the ecCAF signature

The association between the risk score and the clinicopathological characteristics of patients was assessed to expand its applicability value. First, the risk scores of the 5 patients whose tissue samples were collected for single cell sequencing were calculated using the constructed signature, and it was demonstrated that 2 patients with stage III cancer had higher risk scores than the other 3 patients (Table SI). Based on TCGA-CESC dataset, higher risk scores were significantly associated with both a greater number of new tumor events (distant metastasis, locoregional recurrence and new primary tumors; Student's t-test, P<0.05; Fig. 8A) and the tumors were more likely to be incompletely removed by surgery (Student's t-test, P<0.001; Fig. 8B). According to the GSE44001 dataset, patients with higher risk scores tended to have significantly larger tumors (Pearson's correlation coefficient, P<0.001; r=0.189; Fig. 8C; and Wilcoxon rank-sum test, P<0.05; Fig. 8D).

Subsequently, based on TCGA data, clinicopathological characteristics were considered in the construction of a nomogram. A univariate Cox regression model was established to screen possible prognostic factors. A multivariate Cox model was then established to identify independent risk factors affecting the OS rate of patients with cervical cancer. The univariate and multivariate Cox regression results are presented in Table I. It was demonstrated that age (<65 vs. ≥65 years), FIGO stage [early (I–II) vs. late (III–IV)] and risk score (upper vs. lower 50%) were independent risk factors. Based on the Schoenfeld residual plot of proportional hazards test, the P-values of the three variables were all >0.05 (Fig. 8F), which implied that they met the prerequisites of the proportional hazard assumption. This indicated that these variables could be used to establish a nomogram, and the resultant model is presented in Fig. 8E. The nomogram detailed the impact of each factor on CESC and demonstrated that the ecCAF risk score was the greatest influencing factor with respect to the age and stage of cancer of the patient. The C-index was found to be 0.795 (95% confidence interval, 0.743–0.846), which implied that the nomogram had a good discrimination level. The calibration curves of the model demonstrated that the predictive values of the nomogram were highly consistent with the observed values (Fig. 8G), which indicated that the nomogram had a good level of accuracy.

Table I.

Univariate and multivariate Cox proportional hazards regression analysis of the association of clinicopathologic features and overall survival.

Table I.

Univariate and multivariate Cox proportional hazards regression analysis of the association of clinicopathologic features and overall survival.

Cox proportional hazards regression analysis

UnivariateMultivariate


CharacteristicHR (95% CI)P-valueHR (95% CI)P-value
Age (≥65 vs. <65 years)2.049 (1.153–3.641)0.0141.920 (1.081–3.413)0.026
Stage (late vs. early)2.364 (1.426–3.919)0.0012.287 (1.376–3.801)0.001
CAF risk score (upper vs. lower 50%)3.732 (2.157–6.428)0.0003.676 (2.131–6.342)<0.001
Ethnicity
  WhiteReference--0.826
  Black1.127 (0.532–2.387)0.755-0.547
  Asian0.683 (0.165–2.827)0.599-0.488
  Other1.222 (0.615–2.429)0.567-0.739
Histological type
  Squamous carcinomaReference--0.686
  Adenocarcinoma0.949 (0.483–1.865)0.880-0.635
  Adenosquamous carcinoma1.988 (0.271–14.565)0.499-0.481

[i] HR, hazard ratio; CI, confidence interval; CAF, cancer-associated fibroblast.

ecCAF signature is closely associated with tumor-linked pathways

Since the designed ecCAF signature was correlated with adverse prognosis and refractory therapeutic responses, the functional pathways in this model were subsequently assessed using GSEA. Patients in the TCGA-CESC cohort were separated into high- and low-risk groups based on their median ecCAF risk scores. DEGs between the high and low ecCAF score groups were mainly enriched in cancer- and metabolism-associated pathways, including xenobiotic metabolism by cytochrome, oxidative phosphorylation, the TGF-β signaling pathway, and extracellular matrix- and immune-related pathways (P.adjusted <0.05; Fig. 9A; Table SII). Notably, the majority of the signaling pathways were enriched in the high ecCAF score group, whereas the immune-associated pathways were more enriched in the low ecCAF score group. The results obtained using hallmark gene sets were found to be similar; cancer-associated gene sets, including epithelial-mesenchymal transition, hypoxia and TNFA signaling via NFKB, were also significantly enriched in the high ecCAF risk group (P.adjusted<0.05; Fig. 9B).

Role of the ecCAF-based signature in other HPV-associated tumors

In aforementioned results (Fig. 3C and D), KEGG enrichment analysis demonstrated that the HPV infection pathway was enriched in ecCAFs. In addition to cervical cancer, HPV infection has been proposed to contribute to the emergence of a series of different cancer types (55), including HNSC (56), bladder urothelial carcinoma (57), COAD (58) and prostate adenocarcinoma (59). Therefore, a pan-cancer analysis was performed to assess whether the ecCAF-based signature was effective in predicting the prognosis of other HPV-associated cancers. As previously, the OS rate was chosen as the primary outcome. The survival analysis demonstrated that the high ecCAF risk group had a significantly worse prognosis in HNSC (P=0.001) (Fig. 9C) and BLCA (P=0.004) (Fig. 9D), when the observation period was 170 months, whereas no significant differences were identified in terms of COAD or PRAD (Fig. S1B).

The predictive effect of the model on immunotherapy was also evaluated in terms of the four HPV-associated cancers (namely HNSC, COAD, BLCA and PRAD). As demonstrated in Fig. 9E, there were significantly more non-responders in the HNSC high ecCAF risk group compared with the low-risk group, which implied unfavorable effects resulting from therapy (P<0.05); however, for BLCA, COAD and PRAD, there were no differences between the two different risk groups (Fig. S1C).

In terms of chemotherapy, several different chemotherapeutic drugs that are frequently used to treat the four types of HPV-associated cancers were selected for analysis (56,6062), and significant differences were identified between the low and high ecCAF risk score groups in the HNSC, COAD and PRAD cohorts (Wilcoxon rank-sum test, all P<0.05; Fig. 9F-I). Specifically, patients in the high-risk group had a worse chemotherapeutic response compared with the low-risk group, which indicated that the model had potential in terms of its wider applicability. However, the difference in the BLCA cohort was not found to be significant.

Discussion

Cervical cancer is the most common gynecological cancer worldwide and it remains a major health problem among women (63). As reported by the World Health Organization (WHO), 341,831 women died from cervical cancer in 2020, accounting for 7.7% of the total number of cancer-related deaths in women worldwide (1). Currently, first-line treatments after initial diagnosis of the disease are typically surgery or a combination of chemotherapy and radiation, depending on the stage of cancer and other clinicopathological risk factors of the patient (64). However, as one of the vital components in cervical cancer therapy, chemotherapy rarely proves to be curative; moreover, it is associated with a number of adverse side effects and a narrow therapeutic window (65). For example, first-line cisplatin-based chemotherapy yields only a 13% response rate when cisplatin alone is administered, whereas the response rate is 36% for cisplatin double therapy (66,67). The development of effective treatment therapies for cervical cancer, especially advanced cervical cancer, is therefore urgently required.

As an important component of the solid TME, CAFs have long been considered as a potential therapeutic target for tumors. However, numerous therapeutic strategies for CAFs have failed in the clinic, and research in this field of cervical cancer is comparatively lacking. One important reason for this is that CAFs are quite heterogeneous, and the composition and function of CAFs in different types of tumors remain poorly understood (68). However, analysis based on single-cell sequencing may effectively solve these problems. According to the scRNA-seq results of the present study, the composition of CAFs in the cervical cancer microenvironment was relatively simple, and the tumor-promoting effects of ecCAFs were clearly observed. Furthermore, the present study demonstrated the pro-tumorigenic function of ecCAFs using data from additional databases, and this effect was demonstrated to be correlated with several clinical indicators that may be useful in terms of predicting patient prognosis and treatment response.

According to the WHO and the Centers for Disease Control and Prevention, HPV is the second most common carcinogenic infectious agent, ranked only after Helicobacter pylori. HPV induces 31.1% of all cases of infection-caused cancers (69) and ~5% of all cases of cancer (9). As ecCAFs are associated with HPV infection and appear to co-occur in several HPV-associated tumors, it is possible to discuss the role of HPV infection in shaping the tumor stroma. According to a previous study (70), bidirectional crosstalk occurs between HPV-infected epithelial cells and the constituents of the microenvironment. HPV+ cells influence the production of a laminin-rich matrix and are also associated with reduced levels of fibronectin and collagen in fibroblasts (71), increasing the expression levels of pro-angiogenic genes in cells within the adjacent stroma (72) and the direct expression of pro-inflammatory genes in fibroblasts or other stromal cell types (73). Conversely, stromal cells affect the proliferation and differentiation of HPV-infected epithelial cells, and they were reported to serve a key role in an HPV-associated cancer xenograft model (74). The mechanism responsible for crosstalk may be reciprocal secretion and uptake of extracellular vesicles through exosomes. Either the HPV E6 and E7 transcripts or proteins were shown to be transferred to cells via microvesicles (75). These findings not only provide a potential theoretical basis for epithelial-stromal crosstalk, but also represent a major paradigm shift in terms of the understanding of HPV-associated diseases. From this perspective, stromal cells may have certain commonality among different types of HPV-associated cancer, which may prove to be helpful in terms of understanding and controlling HPV-associated cancer.

Moreover, it has been suggested that the ECM may participate in preventing patients from benefiting from the available immunotherapeutic treatments via the exclusion of immune cells. When the tumor stroma is rich in collagen, fibronectin and several proteoglycans (for example, hyaluronic acid and versican), it will have a ‘trapping’ effect on T cells, resulting in inhibition of T cell motility (76). The protease-independent nature of T-cell migration typically results in their movement along the path of least resistance of collagen fibers (77). This is consistent with the findings of the present study where patients with higher ecCAF risk scores tended to have a lower level of immune cell infiltration and were consequently less likely to benefit from immunotherapy. Therefore, targeting ecCAFs in cervical cancer and other HPV-associated tumors may not only have direct antitumor effects, but may also enhance the response rate of patients to immunotherapy. However, this merits further study.

Subsequently, the function of model genes in cancer was assessed in the present study to provide indicators for further research. Although the level of research on cervical cancer remains comparatively deficient, the genes in question have been reported to fulfill roles in other types of cancer. Regarding the eight genes in the signature that were positively correlated with a poor prognosis in the present study, HSPG2 is an extracellular proteoglycan that orchestrates prostate cancer angiogenesis, proliferation, differentiation and invasion (78). Increased levels of HSPG2 expression may therefore be used to independently predict poor OS in neurological tumors and leukemia (79,80). Both LEPROT and LEPR are upregulated in breast cancer tumor tissues, and LEPR has previously been reported to be associated with poor prognosis in breast cancer (81). LEPROT may communicate with the TME, thereby regulating inflammatory or immune signals, and has been reported both to affect cancer development and to serve as a potential prognostic marker or a therapeutic target in pan-cancer (82). In numerous types of cancer, such as lung and prostate cancer, PTGS2 is secreted by CAFs, macrophage type 2 cells and tumor cells, and is therefore reported to have pleiotropic and multifaceted roles in terms of both carcinogenesis and cancer cell resistance to chemotherapy and radiotherapy (83). In liver and colorectal cancer, TMEM9 was reported to hyperactivate Wnt signaling in tumorigenesis through the lysosomal degradation of APC (84). As a component of basement membranes, COL4A1 has been reported to function as a therapeutic target for cancers (85) due to its interaction with other ECM components, thereby participating in epithelial-to-mesenchymal transition, and serving widely pro-tumorigenic roles in different types of cancer. EGLN3 regulates tumor cell apoptosis and proliferation in glioma (86). SEC23A has been reported to function either as an oncogene or as a tumor suppressor gene in different types of cancer and is a potential biomarker for the therapeutic efficacy of docetaxel and vandetanib (87,88). As for the four protective genes (LMO4, VCAM1, RGS5 and NDRG2) in the model constructed in the present study, NDRG2 is widely recognized as a tumor suppressor in several types of cancer (89), whereas the functions of the other three genes remain controversial. RGS5, which co-localizes with platelet/endothelial cell adhesion molecule-1/CD31, platelet derived growth factor receptor-β or α-SMA, typically supports a pro-angiogenic microenvironment (90). Although research in this area is largely lacking, VCAM1 secreted from CAFs was reported in a study to enhance the growth and invasion of lung cancer cells through AKT and MAPK signaling (91). Finally, LMO4 has been reported to participate in tumor proliferation and invasion in numerous types of cancer, and it was also reported to mediate trastuzumab resistance in breast cancer (92).

It should be noted that there were both innovative aspects and limitations associated with the present study. The novelty of the present study is as follows: First, the heterogeneity of CAFs in the cervical cancer microenvironment were assessed and a new subgroup was defined, which was termed ‘extracellular CAF’ on the basis of the scRNA-seq data. As an important component of the cervical cancer microenvironment, ecCAFs have a notable tumor promoting effect. Second, based on the results of the present study, ecCAFs may be prevalent stroma cells in several HPV-associated tumors and directly related to patient prognosis. This suggests the potential value of ecCAFs as a therapeutic target for HPV-associated tumors, which warrants further study. However, it should be acknowledged that the present study has certain limitations: First, the sample size for scRNA-seq analysis was relatively small as it was restricted by research conditions such as lack of funding. Second, the present study preliminarily assessed the function of ecCAFs based on sequencing and public database data, which requires further verification through in vitro and in vivo experiments.

In conclusion, based on the single-cell sequencing of clinical samples, the present study demonstrated the heterogeneity of fibroblasts in the cervical cancer microenvironment, and, to the best of our knowledge, is the first study to identify a new subgroup of CAFs that may be closely associated with tumor progression. A prognostic signature was developed according to the hub genes of ecCAFs, and its clinical significance was analyzed. ecCAFs may be a potential therapeutic target for cervical cancer and other HPV-associated cancers, and these findings may assist in guiding clinical practice and providing the direction of subsequent research.

Supplementary Material

Supporting Data
Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Mr. Zhuangchou Han (from the laboratory of LC-Bio Technologies Hangzhou Co., Ltd.) for their support with single-cell sequencing and analyzing data.

Funding

The present work was supported by the Key Research and Development Plan in Zhejiang Province (grant no. N0.2020C03025), Natural Science Foundation of Zhejiang Province (grant no. LGF21H180011) and Zhejiang Provincial Educational Project (grant no. Y202045540).

Availability of data and materials

The raw sequence data from the 5 patients generated in the present study may be found in the Genome Sequence Archive (Genomics, Proteomics & Bioinformatics 2021) in National Genomics Data Center (Nucleic Acids Res 2022), China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences under accession number, HRA004457, or at the following link https://ngdc.cncb.ac.cn/gsa-human/browse/HRA004457 (93,94). All other data generated in the present study may be requested from the corresponding author.

Authors' contributions

MX collected the clinical samples. YW and YF supervised the single-cell sequencing process and performed preliminary analysis of the raw data. YW and YF confirm the authenticity of all the raw data. YW used the software to complete the main analysis process and wrote the original draft. YY and MX helped with data analysis and interpretation. The fundings was provided by YF, YY and XW. SZ and YL were responsible for validating the results, and supervising the research, and took part in designing the structure of the article. XW and FY provided the conception of the article and reviewed the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was performed according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board (Ethics Committee) of Women's Hospital, School of Medicine, Zhejiang University (Hangzhou, China; approval no. IRB 20200006 R; 2020-01-19). Written informed consent was obtained from the patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TCGA

The Cancer Genome Atlas

CAF

cancer-associated fibroblast

HPA

Human Protein Atlas

TME

tumor microenvironment

FGF

fibroblast growth factor

OS

overall survival

LASSO

least absolute shrinkage and selection operator

TIDE

Tumor Immune Dysfunction and Exclusion

GDSC

Genomics of Drug Sensitivity in Cancer

ROC

receiver operating characteristic

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Cancer Genome Atlas Research Network: Albert Einstein College of Medicine; Analytical Biological Services; Barretos Cancer Hospital; Baylor College of Medicine; Beckman Research Institute of City of Hope; Buck Institute for Research on Aging; Canada's Michael Smith Genome Sciences Centre; Harvard Medical School, . Helen F, et al: Integrated genomic and molecular characterization of cervical cancer. Nature. 543:378–384. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Tewari KS and Monk BJ: Gynecologic oncology group trials of chemotherapy for metastatic and recurrent cervical cancer. Curr Oncol Rep. 7:419–434. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Liontos M, Kyriazoglou A, Dimitriadis I, Dimopoulos MA and Bamias A: Systemic therapy in cervical cancer: 30 Years in review. Crit Rev Oncol Hematol. 137:9–17. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Mutlu L, Tymon-Rosario J, Harold J and Menderes G: Targeted treatment options for the management of metastatic/persistent and recurrent cervical cancer. Expert Rev Anticancer Ther. 22:633–645. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Li C, Teixeira AF, Zhu HJ and ten Dijke P: Cancer associated-fibroblast-derived exosomes in cancer progression. Mol Cancer. 20:1542021. View Article : Google Scholar : PubMed/NCBI

7 

Liu L, Liu L, Yao HH, Zhu ZQ, Ning ZL and Huang Q: Stromal myofibroblasts are associated with poor prognosis in solid cancers: A meta-analysis of published studies. PLoS One. 11:e01599472016. View Article : Google Scholar : PubMed/NCBI

8 

Lavie D, Ben Shmuel A, Erez N and Scherz Shouval R: Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 3:793–807. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Yuan Y, Cai X, Shen F and Ma F: HPV post-infection microenvironment and cervical cancer. Cancer Lett. 497:243–254. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Wright JD, Matsuo K, Huang Y, Tergas AI, Hou JY, Khoury-Collado F, St Clair CM, Ananth CV, Neugut AI and Hershman DL: Prognostic performance of the 2018 international federation of gynecology and obstetrics cervical cancer staging guidelines. Obstet Gynecol. 134:49–57. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Garré JM, Silva HM, Lafaille JJ and Yang G: CX3CR1+ monocytes modulate learning and learning-dependent dendritic spine remodeling via TNF-α. Nat Med. 23:714–722. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Chen Y, Chen S, Li K, Zhang Y, Huang X, Li T, Wu S, Wang Y, Carey LB and Qian W: Overdosage of balanced protein complexes reduces proliferation rate in aneuploid cells. Cell Syst. 9:129–142.e5. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Diskin B, Adam S, Cassini MF, Sanchez G, Liria M, Aykut B, Buttar C, Li E, Sundberg B, Salas RD, et al: PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat Immunol. 21:442–454. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Cano Gamez E, Soskic B, Roumeliotis TI, So E, Smyth DJ, Baldrighi M, Willé D, Nakic N, Esparza Gordillo J, Larminie CGC, et al: Single-cell transcriptomics identifies an effectorness gradient shaping the response of CD4+ T cells to cytokines. Nat Commun. 11:18012020. View Article : Google Scholar : PubMed/NCBI

15 

Xin Z, Lin M, Hao Z, Chen D, Chen Y, Chen X, Xu X, Li J, Wu D, Chai Y and Wu P: The immune landscape of human thymic epithelial tumors. Nat Commun. 13:54632022. View Article : Google Scholar : PubMed/NCBI

16 

Yang C, Siebert JR, Burns R, Gerbec ZJ, Bonacci B, Rymaszewski A, Rau M, Riese MJ, Rao S, Carlson KS, et al: Heterogeneity of human bone marrow and blood natural killer cells defined by single-cell transcriptome. Nat Commun. 10:39312019. View Article : Google Scholar : PubMed/NCBI

17 

Deng Y, Zheng Y, Li D, Hong Q, Zhang M, Li Q, Fu B, Wu L, Wang X, Shen W, et al: Expression characteristics of interferon-stimulated genes and possible regulatory mechanisms in lupus patients using transcriptomics analyses. EBioMedicine. 70:1034772021. View Article : Google Scholar : PubMed/NCBI

18 

Chen Z, Zhou L, Liu L, Hou Y, Xiong M, Yang Y, Hu J and Chen K: Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat Commun. 11:50772020. View Article : Google Scholar : PubMed/NCBI

19 

He D, Wang D, Lu P, Yang N, Xue Z, Zhu X, Zhang P and Fan G: Single-cell RNA sequencing reveals heterogeneous tumor and immune cell populations in early-stage lung adenocarcinomas harboring EGFR mutations. Oncogene. 40:355–368. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Wagner J, Rapsomaniki MA, Chevrier S, Anzeneder T, Langwieder C, Dykgers A, Rees M, Ramaswamy A, Muenst S, Soysal SD, et al: A single-cell atlas of the tumor and immune ecosystem of human breast cancer. Cell. 177:1330–1345.e18. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Zheng H, Liu H, Ge Y and Wang X: Integrated single-cell and bulk RNA sequencing analysis identifies a cancer associated fibroblast-related signature for predicting prognosis and therapeutic responses in colorectal cancer. Cancer Cell Int. 21:5522021. View Article : Google Scholar : PubMed/NCBI

22 

Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP and Tamayo P: The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1:417–425. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Sergushichev AA: An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. bioRxiv. 0600122016.

24 

Wang Z, Li Z, Zhou K, Wang C, Jiang L, Zhang L, Yang Y, Luo W, Qiao W, Wang G, et al: Deciphering cell lineage specification of human lung adenocarcinoma with single-cell RNA sequencing. Nat Commun. 12:65002021. View Article : Google Scholar : PubMed/NCBI

25 

Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, et al: Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 38:675–678. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Lee YY, Kim TJ, Kim JY, Choi CH, Do IG, Song SY, Sohn I, Jung SH, Bae DS, Lee JW and Kim BG: Genetic profiling to predict recurrence of early cervical cancer. Gynecol Oncol. 131:650–654. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Simon N, Friedman J, Hastie T and Tibshirani R: Regularization paths for Cox's proportional hazards model via coordinate descent. J Stat Softw. 39:1–13. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Yan Y, Zhang M and Xu S and Xu S: Identification of an immune gene expression signature for predicting lung squamous cell carcinoma prognosis. Biomed Res Int. 2020:50249422020. View Article : Google Scholar : PubMed/NCBI

29 

Gong Z, Hong F, Wang H, Zhang X and Chen J: An eight-mRNA signature outperforms the lncRNA-based signature in predicting prognosis of patients with glioblastoma. BMC Med Genet. 21:562020. View Article : Google Scholar : PubMed/NCBI

30 

Yang X, Wu W, Pan Y, Zhou Q, Xu J and Han S: Immune-related genes in tumor-specific CD4+ and CD8+ T cells in colon cancer. BMC Cancer. 20:5852020. View Article : Google Scholar : PubMed/NCBI

31 

Hänzelmann S, Castelo R and Guinney J: GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 14:72013. View Article : Google Scholar : PubMed/NCBI

32 

Wang D, Zhang Y, Wang X, Zhang L and Xu S: Construction and validation of an aging-related gene signature predicting the prognosis of pancreatic cancer. Front Genet. 14:10222652023. View Article : Google Scholar : PubMed/NCBI

33 

Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, Li Z, Traugh N, Bu X, Li B, et al: Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 24:1550–1558. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Arina A, Idel C, Hyjek EM, Alegre ML, Wang Y, Bindokas VP, Weichselbaum RR and Schreiber H: Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci USA. 113:7551–7556. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, et al: Genomics of drug sensitivity in cancer (GDSC): A resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 41:(Database Issue). D955–D961. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Maeser D, Gruener RF and Huang RS: oncoPredict: An R package for predicting in vivo or cancer patient drug response and biomarkers from cell line screening data. Brief Bioinform. 22:bbab2602021. View Article : Google Scholar : PubMed/NCBI

37 

Li Y, Lu S, Lan M, Peng X, Zhang Z and Lang J: A prognostic nomogram integrating novel biomarkers identified by machine learning for cervical squamous cell carcinoma. J Transl Med. 18:2232020. View Article : Google Scholar : PubMed/NCBI

38 

Long J, Chen P, Lin J, Bai Y, Yang X, Bian J, Lin Y, Wang D, Yang X, Zheng Y, et al: DNA methylation-driven genes for constructing diagnostic, prognostic, and recurrence models for hepatocellular carcinoma. Theranostics. 9:7251–7267. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI

40 

Song K, Hao J, Ge Z and Chen P: Clinical and survival impact of sex-determining region Y-Box 2 in colorectal cancer: An integrated analysis of the immunohistochemical study and bioinformatics analysis. J Oncol. 2020:37615352020. View Article : Google Scholar : PubMed/NCBI

41 

Yoshida GJ: Regulation of heterogeneous cancer-associated fibroblasts: The molecular pathology of activated signaling pathways. J Exp Clin Cancer Res. 39:1122020. View Article : Google Scholar : PubMed/NCBI

42 

Au Yeung CL, Tsang TY, Yau PL and Kwok TT: Human papillomavirus type 16 E6 induces cervical cancer cell migration through the p53/microRNA-23b/urokinase-type plasminogen activator pathway. Oncogene. 30:2401–2410. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Sato T, Sakai T, Noguchi Y, Takita M, Hirakawa S and Ito A: Tumor-stromal cell contact promotes invasion of human uterine cervical carcinoma cells by augmenting the expression and activation of stromal matrix metalloproteinases. Gynecol Oncol. 92:47–56. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Zhang W, Ge Y, Cheng Q, Zhang Q, Fang L and Zheng J: Decorin is a pivotal effector in the extracellular matrix and tumour microenvironment. Oncotarget. 9:5480–5491. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Schulz WA, Ingenwerth M, Djuidje CE, Hader C, Rahnenführer J and Engers R: Changes in cortical cytoskeletal and extracellular matrix gene expression in prostate cancer are related to oncogenic ERG deregulation. BMC Cancer. 10:5052010. View Article : Google Scholar : PubMed/NCBI

46 

Lucarelli G, Rutigliano M, Bettocchi C, Palazzo S, Vavallo A, Galleggiante V, Trabucco S, Di Clemente D, Selvaggi FP, Battaglia M and Ditonno P: Spondin-2, a secreted extracellular matrix protein, is a novel diagnostic biomarker for prostate cancer. J Urol. 190:2271–2277. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Kubala JM, Laursen KB, Schreiner R, Williams RM, van der Mijn JC, Crowley MJ, Mongan NP, Nanus DM, Heller DA and Gudas LJ: NDUFA4L2 reduces mitochondrial respiration resulting in defective lysosomal trafficking in clear cell renal cell carcinoma. Cancer Biol Ther. 24:21706692023. View Article : Google Scholar : PubMed/NCBI

48 

Chen PH, Shen WL, Shih CM, Ho KH, Cheng CH, Lin CW, Lee CC, Liu AJ and Chen KC: The CHAC1-inhibited Notch3 pathway is involved in temozolomide-induced glioma cytotoxicity. Neuropharmacology. 116:300–314. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Deaton RA, Bulut G, Serbulea V, Salamon A, Shankman LS, Nguyen AT and Owens GK: A new autosomal Myh11-CreERT2 smooth muscle cell lineage tracing and gene knockout mouse model-brief report. Arterioscler Thromb Vasc Biol. 43:203–211. 2023. View Article : Google Scholar : PubMed/NCBI

50 

Loumaye A, Lause P, Zhong X, Zimmers TA, Bindels LB and Thissen JP: Activin a causes muscle atrophy through MEF2C-dependent impaired myogenesis. Cells. 11:11192022. View Article : Google Scholar : PubMed/NCBI

51 

Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres-Garcia W, Treviño V, Shen H, Laird PW, Levine DA, et al: Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 4:26122013. View Article : Google Scholar : PubMed/NCBI

52 

Chiossone L, Dumas PY, Vienne M and Vivier E: Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol. 18:671–688. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Giles JR, Globig AM, Kaech SM and Wherry EJ: CD8+ T cells in the cancer-immunity cycle. Immunity. 56:2231–2253. 2023. View Article : Google Scholar : PubMed/NCBI

54 

Speiser DE, Chijioke O, Schaeuble K and Münz C: CD4+ T cells in cancer. Nat Cancer. 4:317–329. 2023. View Article : Google Scholar : PubMed/NCBI

55 

Wei E, Reisinger A, Li J, French LE, Clanner-Engelshofen B and Reinholz M: Integration of scRNA-Seq and TCGA RNA-Seq to analyze the heterogeneity of HPV+ and HPV-cervical cancer immune cells and establish molecular risk models. Front Oncol. 12:8609002022. View Article : Google Scholar : PubMed/NCBI

56 

Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 6:922020. View Article : Google Scholar : PubMed/NCBI

57 

Shaker OG, Hammam OA and Wishahi MM: Is there a correlation between HPV and urinary bladder carcinoma? Biomed Pharmacother. 67:183–191. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Pelizzer T, Dias CP, Poeta J, Torriani T and Roncada C: Colorectal cancer prevalence linked to human papillomavirus: A systematic review with meta-analysis. Rev Bras Epidemiol. 19:791–802. 2016.(In Portuguese, English). View Article : Google Scholar : PubMed/NCBI

59 

Chen AC, Waterboer T, Keleher A, Morrison B, Jindal S, McMillan D, Nicol D, Gardiner RA, McMillan NA and Antonsson A: Human papillomavirus in benign prostatic hyperplasia and prostatic adenocarcinoma patients. Pathol Oncol Res. 17:613–617. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Kamat AM, Hahn NM, Efstathiou JA, Lerner SP, Malmström PU, Choi W, Guo CC, Lotan Y and Kassouf W: Bladder cancer. Lancet. 388:2796–2810. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Gelibter AJ, Caponnetto S, Urbano F, Emiliani A, Scagnoli S, Sirgiovanni G, Napoli VM and Cortesi E: Adjuvant chemotherapy in resected colon cancer: When, how and how long? Surg Oncol. 30:100–107. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Schatten H: Brief overview of prostate cancer statistics, grading, diagnosis and treatment strategies. Adv Exp Med Biol. 1095:1–14. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Shanmugaraj B, Malla A, Bulaon CJI, Phoolcharoen W and Phoolcharoen N: Harnessing the potential of plant expression system towards the production of vaccines for the prevention of human papillomavirus and cervical cancer. Vaccines (Basel). 10:20642022. View Article : Google Scholar : PubMed/NCBI

64 

National Comprehensive Cancer Network and Clinical practice guidelines in oncology cervical cancer, . version 4. 2019. https://www.nccn.org/professionals/physician_gls/pdf/cervical.pdfMarch 29–2019.

65 

Mauricio D, Zeybek B, Tymon-Rosario J, Harold J and Santin AD: Immunotherapy in cervical cancer. Curr Oncol Rep. 23:612021. View Article : Google Scholar : PubMed/NCBI

66 

Long HJ III, Bundy BN, Grendys EC Jr, Benda JA, McMeekin DS, Sorosky J, Miller DS, Eaton LA and Fiorica JV; Gynecologic Oncology Group Study, : Randomized phase III trial of cisplatin with or without topotecan in carcinoma of the uterine cervix: A gynecologic oncology group study. J Clin Oncol. 23:4626–4633. 2005. View Article : Google Scholar : PubMed/NCBI

67 

Monk BJ, Sill MW, McMeekin DS, Cohn DE, Ramondetta LM, Boardman CH, Benda J and Cella D: Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: A gynecologic oncology group study. J Clin Oncol. 27:4649–4655. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Chen Y, McAndrews KM and Kalluri R: Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 18:792–804. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Szymonowicz KA and Chen J: Biological and clinical aspects of HPV-related cancers. Cancer Biol Med. 17:864–878. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Spurgeon ME and Lambert PF: Human papillomavirus and the stroma: Bidirectional crosstalk during the virus life cycle and carcinogenesis. Viruses. 9:2192017. View Article : Google Scholar : PubMed/NCBI

71 

Fullár A, Dudás J, Oláh L, Hollósi P, Papp Z, Sobel G, Karászi K, Paku S, Baghy K and Kovalszky I: Remodeling of extracellular matrix by normal and tumor-associated fibroblasts promotes cervical cancer progression. BMC Cancer. 15:2562015. View Article : Google Scholar : PubMed/NCBI

72 

Pilch H, Schlenger K, Steiner E, Brockerhoff P, Knapstein P and Vaupel P: Hypoxia-stimulated expression of angiogenic growth factors in cervical cancer cells and cervical cancer-derived fibroblasts. Int J Gynecol Cancer. 11:137–142. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Woodby B, Scott M and Bodily J: The interaction between human papillomaviruses and the stromal microenvironment. Prog Mol Biol Transl Sci. 144:169–238. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Murata T, Mizushima H, Chinen I, Moribe H, Yagi S, Hoffman RM, Kimura T, Yoshino K, Ueda Y, Enomoto T and Mekada E: HB-EGF and PDGF mediate reciprocal interactions of carcinoma cells with cancer-associated fibroblasts to support progression of uterine cervical cancers. Cancer Res. 71:6633–6642. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Chiantore MV, Mangino G, Iuliano M, Zangrillo MS, De Lillis I, Vaccari G, Accardi R, Tommasino M, Columba Cabezas S, Federico M, et al: Human papillomavirus E6 and E7 oncoproteins affect the expression of cancer-related microRNAs: Additional evidence in HPV induced tumorigenesis. J Cancer Res Clin Oncol. 142:1751–1763. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Evanko SP, Potter-Perigo S, Bollyky PL, Nepom GT and Wight TN: Hyaluronan and versican in the control of human T-lymphocyte adhesion and migration. Matrix Biol. 31:90–100. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Bougherara H, Mansuet-Lupo A, Alifano M, Ngô C, Damotte D, Le Frère-Belda MA, Donnadieu E and Peranzoni E: Real-time imaging of resident T cells in human lung and ovarian carcinomas reveals how different tumor microenvironments control T lymphocyte migration. Front Immunol. 6:5002015. View Article : Google Scholar : PubMed/NCBI

78 

Grindel BJ, Martinez JR, Tellman TV, Harrington DA, Zafar H, Nakhleh L, Chung LW and Farach Carson MC: Matrilysin/MMP-7 cleavage of perlecan/HSPG2 complexed with semaphorin 3A supports FAK-mediated stromal invasion by prostate cancer cells. Sci Rep. 8:72622018. View Article : Google Scholar : PubMed/NCBI

79 

Kazanskaya GM, Tsidulko AY, Volkov AM, Kiselev RS, Suhovskih AV, Kobozev VV, Gaytan AS, Aidagulova SV, Krivoshapkin AL and Grigorieva EV: Heparan sulfate accumulation and perlecan/HSPG2 up-regulation in tumour tissue predict low relapse-free survival for patients with glioblastoma. Histochem Cell Biol. 149:235–244. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Zhou X, Liang S, Zhan Q, Yang L, Chi J and Wang L: HSPG2 overexpression independently predicts poor survival in patients with acute myeloid leukemia. Cell Death Dis. 11:4922020. View Article : Google Scholar : PubMed/NCBI

81 

Li L, Meng X, Liu L, Xiang Y, Wang F, Yu L, Zhou F, Zheng C, Zhou W, Cui S, et al: Single-nucleotide polymorphisms in LEP and LEPR associated with breast cancer risk: Results from a multicenter case-control study in Chinese females. Front Oncol. 12:8095702022. View Article : Google Scholar : PubMed/NCBI

82 

Li B, He Y, Li P and Chen X: Leptin receptor overlapping transcript (LEPROT) is associated with the tumor microenvironment and a prognostic predictor in pan-cancer. Front Genet. 12:7494352021. View Article : Google Scholar : PubMed/NCBI

83 

Hashemi Goradel N, Najafi M, Salehi E, Farhood B and Mortezaee K: Cyclooxygenase-2 in cancer: A review. J Cell Physiol. 234:5683–5699. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Jung YS, Stratton SA, Lee SH, Kim MJ, Jun S, Zhang J, Zheng B, Cervantes CL, Cha JH, Barton MC and Park JI: TMEM9-v-ATPase activates Wnt/β-catenin signaling via APC lysosomal degradation for liver regeneration and tumorigenesis. Hepatology. 73:776–794. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Zhang H, Wang Y and Ding H: COL4A1, negatively regulated by XPD and miR-29a-3p, promotes cell proliferation, migration, invasion and epithelial-mesenchymal transition in liver cancer cells. Clin Transl Oncol. 23:2078–2089. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Mao K, You C, Lei D and Zhang H: Potential regulation of glioma through the induction of apoptosis signaling via Egl-9 family hypoxia-inducible factor 3. Oncol Lett. 13:893–897. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Wang Y, Lieberman R, Pan J, Zhang Q, Du M, Zhang P, Nevalainen M, Kohli M, Shenoy NK, Meng H, et al: miR-375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. Mol Cancer. 15:702016. View Article : Google Scholar : PubMed/NCBI

88 

Lassalle S, Zangari J, Popa A, Ilie M, Hofman V, Long E, Patey M, Tissier F, Belléannée G, Trouette H, et al: MicroRNA-375/SEC23A as biomarkers of the in vitro efficacy of vandetanib. Oncotarget. 7:30461304782016. View Article : Google Scholar : PubMed/NCBI

89 

Chen XL, Lei L, Hong LL and Ling ZQ: Potential role of NDRG2 in reprogramming cancer metabolism and epithelial-to-mesenchymal transition. Histol Histopathol. 33:655–663. 2018.PubMed/NCBI

90 

Silini A, Ghilardi C, Figini S, Sangalli F, Fruscio R, Dahse R, Pedley RB, Giavazzi R and Bani M: Regulator of G-protein signaling 5 (RGS5) protein: A novel marker of cancer vasculature elicited and sustained by the tumor's proangiogenic microenvironment. Cell Mol Life Sci. 69:1167–1178. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Zhou Z, Zhou Q, Wu X, Xu S, Hu X, Tao X, Li B, Peng J, Li D, Shen L, et al: VCAM-1 secreted from cancer-associated fibroblasts enhances the growth and invasion of lung cancer cells through AKT and MAPK signaling. Cancer Lett. 473:62–73. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Ding K, Wu Z, Li X, Sheng Y, Wang X and Tan S: LMO4 mediates trastuzumab resistance in HER2 positive breast cancer cells. Am J Cancer Res. 8:594–609. 2018.PubMed/NCBI

93 

Chen T, Chen X, Zhang S, Zhu J, Tang B, Wang A, Dong L, Zhang Z, Yu C, Sun Y, et al: The genome sequence archive family: toward explosive data growth and diverse data types. Genomics Proteomics Bioinformatics. 19:578–583. 2021. View Article : Google Scholar : PubMed/NCBI

94 

CNCB-NGDC Members, . Partners Database resources of the national genomics data center, China national center for bioinformation in 2022. Nucleic Acids Res. 50(D1): D27–D38. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Xu M, Yao Y, Li Y, Zhang S, Fu Y and Wang X: Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions. Oncol Lett 27: 167, 2024
APA
Wang, Y., Xu, M., Yao, Y., Li, Y., Zhang, S., Fu, Y., & Wang, X. (2024). Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions. Oncology Letters, 27, 167. https://doi.org/10.3892/ol.2024.14300
MLA
Wang, Y., Xu, M., Yao, Y., Li, Y., Zhang, S., Fu, Y., Wang, X."Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions". Oncology Letters 27.4 (2024): 167.
Chicago
Wang, Y., Xu, M., Yao, Y., Li, Y., Zhang, S., Fu, Y., Wang, X."Extracellular cancer‑associated fibroblasts: A novel subgroup in the cervical cancer microenvironment that exhibits tumor‑promoting roles and prognosis biomarker functions". Oncology Letters 27, no. 4 (2024): 167. https://doi.org/10.3892/ol.2024.14300